201
|
Dressman J, Kincer J, Matveev SV, Guo L, Greenberg RN, Guerin T, Meade D, Li XA, Zhu W, Uittenbogaard A, Wilson ME, Smart EJ. HIV protease inhibitors promote atherosclerotic lesion formation independent of dyslipidemia by increasing CD36-dependent cholesteryl ester accumulation in macrophages. J Clin Invest 2003. [DOI: 10.1172/jci200316261] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
202
|
Dressman J, Kincer J, Matveev SV, Guo L, Greenberg RN, Guerin T, Meade D, Li XA, Zhu W, Uittenbogaard A, Wilson ME, Smart EJ. HIV protease inhibitors promote atherosclerotic lesion formation independent of dyslipidemia by increasing CD36-dependent cholesteryl ester accumulation in macrophages. J Clin Invest 2003; 111:389-97. [PMID: 12569165 PMCID: PMC151854 DOI: 10.1172/jci16261] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Protease inhibitors decrease the viral load in HIV patients, however the patients develop hypertriglyceridemia, hypercholesterolemia, and atherosclerosis. It has been assumed that protease inhibitor-dependent increases in atherosclerosis are secondary to the dyslipidemia. Incubation of THP-1 cells or human PBMCs with protease inhibitors caused upregulation of CD36 and the accumulation of cholesteryl esters. The use of CD36-blocking antibodies, a CD36 morpholino, and monocytes isolated from CD36 null mice demonstrated that protease inhibitor-induced increases in cholesteryl esters were dependent on CD36 upregulation. These data led to the hypothesis that protease inhibitors induce foam cell formation and consequently atherosclerosis by upregulating CD36 and cholesteryl ester accumulation independent of dyslipidemia. Studies with LDL receptor null mice demonstrated that low doses of protease inhibitors induce an increase in the level of CD36 and cholesteryl ester in peritoneal macrophages and the development of atherosclerosis without altering plasma lipids. Furthermore, the lack of CD36 protected the animals from protease inhibitor-induced atherosclerosis. Finally, ritonavir increased PPAR-gamma and CD36 mRNA levels in a PKC- and PPAR-gamma-dependent manner. We conclude that protease inhibitors contribute to the formation of atherosclerosis by promoting the upregulation of CD36 and the subsequent accumulation of sterol in macrophages.
Collapse
Affiliation(s)
- James Dressman
- Department of Physiology, University of Kentucky Medical School, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Mann GE, Yudilevich DL, Sobrevia L. Regulation of amino acid and glucose transporters in endothelial and smooth muscle cells. Physiol Rev 2003; 83:183-252. [PMID: 12506130 DOI: 10.1152/physrev.00022.2002] [Citation(s) in RCA: 284] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
While transport processes for amino acids and glucose have long been known to be expressed in the luminal and abluminal membranes of the endothelium comprising the blood-brain and blood-retinal barriers, it is only within the last decades that endothelial and smooth muscle cells derived from peripheral vascular beds have been recognized to rapidly transport and metabolize these nutrients. This review focuses principally on the mechanisms regulating amino acid and glucose transporters in vascular endothelial cells, although we also summarize recent advances in the understanding of the mechanisms controlling membrane transport activity and expression in vascular smooth muscle cells. We compare the specificity, ionic dependence, and kinetic properties of amino acid and glucose transport systems identified in endothelial cells derived from cerebral, retinal, and peripheral vascular beds and review the regulation of transport by vasoactive agonists, nitric oxide (NO), substrate deprivation, hypoxia, hyperglycemia, diabetes, insulin, steroid hormones, and development. In view of the importance of NO as a modulator of vascular tone under basal conditions and in disease and chronic inflammation, we critically review the evidence that transport of L-arginine and glucose in endothelial and smooth muscle cells is modulated by bacterial endotoxin, proinflammatory cytokines, and atherogenic lipids. The recent colocalization of the cationic amino acid transporter CAT-1 (system y(+)), nitric oxide synthase (eNOS), and caveolin-1 in endothelial plasmalemmal caveolae provides a novel mechanism for the regulation of NO production by L-arginine delivery and circulating hormones such insulin and 17beta-estradiol.
Collapse
Affiliation(s)
- Giovanni E Mann
- Centre for Cardiovascular Biology and Medicine, Guy's, King's, and St. Thomas' School of Biomedical Sciences, King's College London, London, United Kingdom.
| | | | | |
Collapse
|
204
|
Wratten ML, Galaris D, Tetta C, Sevanian A. Evolution of oxidative stress and inflammation during hemodialysis and their contribution to cardiovascular disease. Antioxid Redox Signal 2002; 4:935-44. [PMID: 12573142 DOI: 10.1089/152308602762197470] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
End-stage renal disease patients have increased cardiovascular morbidity and mortality. These patients have many unique risk factors, such as an accumulation of uremic toxins, electrolyte imbalances, metabolic disturbances, anemia, chronic inflammation, and thrombogenic disturbances. Oxidative stress has been implicated in many of these disturbances. This review will focus on some of the factors that may accelerate cardiovascular disease in uremic patients, with an emphasis on mechanisms and interactions of various components of oxidative stress and inflammation. Understanding the mechanisms of these pathways may be useful in developing effective prevention and treatment strategies.
Collapse
Affiliation(s)
- Mary Lou Wratten
- Clinical and Laboratory Research Department, Bellco, Mirandola (MO) Italy.
| | | | | | | |
Collapse
|
205
|
Abstract
Accumulating evidence has suggested the protective role of HDL in cardiovascular disease processes. Calcification is a common feature of atherosclerotic lesions and contributes to cardiovascular complications due to the loss of aortic resilience and function. Recent studies have suggested that vascular calcification shares several features with skeletal bone formation at the cellular and molecular levels. These include the presence of osteoblast-like calcifying vascular cells in the artery wall that undergo osteoblastic differentiation and calcification in vitro. We hypothesized that HDL may also protect against vascular calcification by regulating the osteogenic activity of these calcifying vascular cells. When treated with HDL, alkaline phosphatase activity, a marker of osteogenic differentiation of osteoblastic cells, was significantly reduced in those cells. Prolonged treatment with HDL also inhibited calcification of these cells, further supporting the antiosteogenic differentiation property of HDL when applied to vascular cells. Furthermore, HDL inhibited the osteogenic activity that was induced by inflammatory cytokines interleukin (IL)-1beta and IL-6 as well as by minimally oxidized LDL. HDL also partially inhibited the IL-6-induced activation of signal transducer and activator of transcription 3 in calcifying vascular cells, suggesting that HDL may inhibit cytokine-induced signal transduction pathways. The inhibitory effects of HDL were mimicked by lipids extracted from HDL but not by HDL-associated apolipoproteins or reconstituted HDL. Furthermore, oxidation of HDL rendered it pro-osteogenic. Taken together, these results suggest that HDL regulates the osteoblastic differentiation and calcification of vascular cells and that vascular calcification may be another target of HDL action in the artery wall.
Collapse
Affiliation(s)
- Farhad Parhami
- Department of Medicine, University of California, Los Angeles 90095, USA.
| | | | | | | | | |
Collapse
|
206
|
Gouni-Berthold I, Sachinidis A. Does the coronary risk factor low density lipoprotein alter growth and signaling in vascular smooth muscle cells? FASEB J 2002; 16:1477-87. [PMID: 12374770 DOI: 10.1096/fj.02-0260rev] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
There is increasing evidence that hypertension promotes low density lipoprotein (LDL) transportation into the subendothelial space of the vascular wall. Vascular smooth muscle cell (VSMC) proliferation plays an important role in the development and progression of cardiovascular diseases. Recently, several studies have demonstrated that LDL acts as a classic growth factor promoting VSMC growth via mitogenic signals normally elicited by classic growth factors. The present work summarizes current nontraditional concepts regarding possible cellular mechanisms through which hypertension and LDL may promote the development of atherosclerosis. Especially addressed are the possible effects of an elevated blood pressure in combination with LDL on VSMC growth. The new research concept concerning LDL as a growth factor and carrier for biological active phospholipids such as sphingosine-1-phosphate and sphingosylphosphorylcholine may contribute to an understanding of the pathogenesis of atherosclerosis by elevated high blood pressure.
Collapse
|
207
|
Silver DL. A carboxyl-terminal PDZ-interacting domain of scavenger receptor B, type I is essential for cell surface expression in liver. J Biol Chem 2002; 277:34042-7. [PMID: 12119305 DOI: 10.1074/jbc.m206584200] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Scavenger receptor B, type I (SR-BI) was recently shown to interact with a PDZ domain-containing protein, PDZK1 (CLAMP/Diphor-1/CAP70/NaPi-Cap1), but the importance of this interaction in vivo in terms of SR-BI function has not been determined. In an effort to elucidate the role of this interaction in vivo, the PDZK1-interacting domain of SR-BI was identified and mutated and expressed liver-specifically in mice. The PDZKI-interacting domain on SR-BI was identified as the last three carboxyl-terminal amino acids, Arg-Lys-Leu. A mutant SR-BI (SR-BIdel509) that lacked only the leucine in the PDZ-interacting domain failed to interact with PDZK1 in vitro, while showing normal selective uptake function in nonpolarized cells. Transgenic mice with liver overexpression of SR-BIdel509 showed marked accumulation of SR-BI mRNA with only a moderate increase in SR-BI protein in liver, with no reduction in plasma cholesterol levels. Measurement of cell surface SR-BI levels and HDL cholesteryl ester-selective uptake in primary hepatocytes from transgenic mice revealed that SR-BIdel509 was not expressed at the plasma membrane correlating with normal levels of selective uptake compared with hepatocytes from nontransgenic littermates. This study indicates that the PDZK1-interacting domain of SR-BI is essential for cell surface expression of SR-BI in liver and suggests that PDZK1 or other PDZ domain proteins may play an important role in regulating SR-BI cell surface expression and hence reverse cholesterol transport.
Collapse
Affiliation(s)
- David L Silver
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
208
|
Smart EJ, Anderson RGW. Alterations in membrane cholesterol that affect structure and function of caveolae. Methods Enzymol 2002; 353:131-9. [PMID: 12078489 DOI: 10.1016/s0076-6879(02)53043-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Most of the available methods for modifying caveolae structure and function depend on altering the cholesterol content of caveolae. The most important aspect of each method is to ensure the reagents are working in the cells that are being studied. The idiosyncrasies of each method are such that they cannot be universally applied without carefully optimizing the conditions. When used correctly, these methods are accepted as a specific way to perturb the structure and function of caveolae.
Collapse
Affiliation(s)
- Eric J Smart
- Department of Physiology, University of Kentucky Medical School, Lexington, Kentucky 40536, USA
| | | |
Collapse
|
209
|
Li D, Chen H, Romeo F, Sawamura T, Saldeen T, Mehta JL. Statins modulate oxidized low-density lipoprotein-mediated adhesion molecule expression in human coronary artery endothelial cells: role of LOX-1. J Pharmacol Exp Ther 2002; 302:601-5. [PMID: 12130721 DOI: 10.1124/jpet.102.034959] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
LOX-1, a receptor for oxidized low-density lipoprotein (ox-LDL), plays a critical role in endothelial dysfunction and atherosclerosis. LOX-1 activation also plays an important role in monocyte adhesion to endothelial cells. A number of studies show that 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) reduce total LDL cholesterol and exert a cardioprotective effect. We examined the modulation of LOX-1 expression and its function by two different statins, simvastatin and atorvastatin, in human coronary artery endothelial cells (HCAECs). We observed that ox-LDL (40 microg/ml) treatment up-regulated the expression of E- and P-selectins, VCAM-1 and ICAM-1 in HCAECs. Ox-LDL mediated these effects via LOX-1, since antisense to LOX-1 mRNA decreased LOX-1 expression and subsequent adhesion molecule expression. Pretreatment of HCAECs with simvastatin or atorvastatin (1 and 10 microM) reduced ox-LDL-induced expression of LOX-1 as well as adhesion molecules (all P < 0.05). A high concentration of statins (10 microM) was more potent than the low concentration (1 microM) (P < 0.05). Both statins reduced ox-LDL-mediated activation of the redox-sensitive nuclear factor-kappaB (NF-kappaB) but not AP-1. These observations indicate that LOX-1 activation plays an important role in ox-LDL-induced expression of adhesion molecules. Inhibition of expression of LOX-1 and adhesion molecules and activation of NF-kappaB may be another mechanism of beneficial effects of statins in vascular diseases.
Collapse
Affiliation(s)
- Dayuan Li
- Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 W Markham, Little Rock, AR 72205, USA
| | | | | | | | | | | |
Collapse
|
210
|
Kincer JF, Uittenbogaard A, Dressman J, Guerin TM, Febbraio M, Guo L, Smart EJ. Hypercholesterolemia promotes a CD36-dependent and endothelial nitric-oxide synthase-mediated vascular dysfunction. J Biol Chem 2002; 277:23525-33. [PMID: 11976335 DOI: 10.1074/jbc.m202465200] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Numerous studies have implicated either the presence or absence of CD36 in the development of hypertension. In addition, hypercholesterolemia is associated with the loss of nitric oxide-induced vasodilation and the subsequent increase in blood pressure. In the current study, we tested the hypothesis that diet-induced hypercholesterolemia promotes the disruption of agonist-stimulated nitric oxide generation and vasodilation in a CD36-dependent manner. To test this, C57BL/6, apoE null, CD36 null, and apoE/CD36 null mice were maintained on chow or high fat diets. In contrast to apoE null mice fed a chow diet, apoE null mice fed a high fat diet did not respond to acetylcholine with a decrease in blood pressure. Caveolae isolated from in vivo vessels did not contain endothelial nitric-oxide synthase and were depleted of cholesterol. Age-matched apoE/CD36 null mice fed a chow or high fat diet responded to acetylcholine with a decrease in blood pressure. The mechanism underlying the vascular dysfunction was reversible because vessels isolated from apoE null high fat-fed mice regained responsiveness to acetylcholine when incubated with plasma obtained from chow-fed mice. Further analysis demonstrated that the plasma low density lipoprotein fraction was responsible for depleting caveolae of cholesterol, removing endothelial nitric-oxide synthase from caveolae, and preventing nitric oxide production. In addition, the pharmacological removal of caveola cholesterol with cyclodextrin mimicked the effects caused by the low density lipoprotein fraction. We conclude that the ablation of CD36 prevented the negative impact of hypercholesterolemia on agonist-stimulated nitric oxide-mediated vasodilation in apoE null mice. These studies provide a direct link between CD36 and the early events that underlie hypercholesterolemia-mediated hypertension and mechanistic linkages between CD36 function, nitric-oxide synthase activation, caveolae integrity, and blood pressure regulation.
Collapse
Affiliation(s)
- Jeanie F Kincer
- University of Kentucky Medical School, Department of Physiology, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | |
Collapse
|
211
|
Wagner S, Groschner K, Mayer B, Schmidt K. Desensitization of endothelial nitric oxide synthase by receptor agonists. Biochem J 2002; 364:863-8. [PMID: 12049652 PMCID: PMC1222637 DOI: 10.1042/bj20011178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Stimulation of endothelial cells with receptor agonists leads to an activation of endothelial nitric oxide synthase (eNOS) that only lasts for a short duration. A more prolonged effect, however, is observed in response to non-receptor agonists, such as Ca2+ ionophores or thapsigargin (TG). To investigate the molecular mechanisms underlying the rapid deactivation of eNOS after stimulation with receptor agonists, we measured the time courses of eNOS activation and intracellular free Ca2+ concentration ([Ca2+]i) in response to bradykinin (BK) and ATP. Incubation of porcine aortic endothelial cells with BK (1 microM) in the presence of 3 mM extracellular Ca2+ increased [Ca2+]i from 110 to 350 nM and enhanced the rate of l-[3H]citrulline formation from 0.1 to 5 fmol/min. In the absence of extracellular Ca2+, the BK-induced increase in [Ca2+]i was only marginal (from 30 to 110 nM) and not sufficient to activate eNOS. When Ca(2+) (final concentration 3 mM) was added 10 min after BK, [Ca2+]i increased to 330 nM within 3 min, but interestingly, formation of l-[3H]citrulline was not detectable. A similar phenomenon was observed with ATP, but not with Ca2+ ionophores or TG. This indicates that stimulation of endothelial cells with receptor agonists leads to desensitization of eNOS, which renders the enzyme insensitive to activation by subsequent increases in [Ca2+]i. However, when ATP was added to BK-pretreated cells or, conversely, BK to ATP-pretreated cells, activation of eNOS was comparable with that of untreated cells, suggesting that BK and ATP affect different pools of eNOS. The desensitization of eNOS was reversible, since removal of ATP or BK from the incubation buffer restored the response to the respective agonist within 20 min. In addition to the transient Ca2+ signal, desensitization of eNOS may represent a further mechanism by which endothelial cells rapidly terminate receptor-dependent NO formation.
Collapse
Affiliation(s)
- Sabine Wagner
- Institut für Pharmakologie und Toxikologie, Karl-Franzens-Universität Graz, Universitätsplatz 2, A-8010 Graz, Austria
| | | | | | | |
Collapse
|
212
|
Abstract
Caveolae are spherical invaginations of the plasma membrane and associated vesicles that are found at high surface densities in most cells, endothelia included. Their structural framework has been shown to consist of oligomerized caveolin molecules interacting with cholesterol and sphingolipids. Caveolae have been involved in many cellular functions such as endocytosis, signal transduction, mechano-transduction, potocytosis, and cholesterol trafficking. Some confusion still persists in the field with respect to the relationship between caveolae and the lipid rafts, which have been involved in many of the above functions. In addition to all these, endothelial caveolae have been involved in capillary permeability by their participation in the process of transcytosis. This short review will focus on their structure and components, methods used to determine these components, and the role of caveolae in the transendothelial exchanges between blood plasma and the interstitial fluid.
Collapse
Affiliation(s)
- Radu-Virgil Stan
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California 92093-0651, USA.
| |
Collapse
|
213
|
Abstract
Endothelial nitric oxide synthase (eNOS) is expressed in vascular endothelium, airway epithelium, and certain other cell types where it generates the key signaling molecule nitric oxide (NO). Diminished NO availability contributes to systemic and pulmonary hypertension, atherosclerosis, and airway dysfunction. Complex mechanisms underly the cell specificity of eNOS expression, and co- and post-translational processing leads to trafficking of the enzyme to plasma membrane caveolae. Within caveolae, eNOS is the downstream target member of a signaling complex in which it is functionally linked to both typical G protein-coupled receptors and less typical receptors such as estrogen receptor (ER) alpha and the high-density lipoprotein receptor SR-BI displaying novel actions. This compartmentalization facilitates dynamic protein-protein interactions and calcium- and phosphorylation-dependent signal transduction events that modify eNOS activity. Further understanding of these mechanisms will enable us to take preventive and therapeutic advantage of the powerful actions of NO in multiple cell types.
Collapse
Affiliation(s)
- Philip W Shaul
- Department of Pediatrics University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9063, USA.
| |
Collapse
|
214
|
Abstract
Accelerated atherosclerosis is often observed in patients with chronic renal failure. In the present review we summarize and discuss the recent literature on the pathogenic role of low-density lipoproteins modified by oxidative processes in atherosclerosis and the possible role in renal diseases. Pathogenetically, the oxidation of low-density lipoproteins is considered to be a key event in the development of atherosclerosis, in part by causing enhanced uptake of lipids by macrophages. In addition, oxidation of low-density lipoproteins exerts cytotoxic, proinflammatory and immunogenic properties, all of which could potentially contribute to the development and progression of atherosclerosis.
Collapse
Affiliation(s)
- Peter Heeringa
- Department of Clinical and Experimental Immunology, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands.
| | | |
Collapse
|
215
|
Abstract
This review considers the antiatherogenic function of high density lipoprotein (HDL) from the point of view of its dynamics within the sequential steps of reverse cholesterol transport (RCT). It is postulated that the efficiency of cholesterol flux through the RCT pathways is clinically more relevant than the HDL cholesterol concentration. The particular role of pre-beta(1)-HDL is reviewed drawing attention to the relationship between its concentration and the flux of cholesterol through the RCT system.
Collapse
Affiliation(s)
- Dmitri Sviridov
- Baker Medical Research Institute, PO Box 6492, St. Kilda Rd. Central, Melbourne, Vic. 8008, Australia.
| | | |
Collapse
|
216
|
Broeders MAW, Tangelder GJ, Slaaf DW, Reneman RS, oude Egbrink MGA. Hypercholesterolemia enhances thromboembolism in arterioles but not venules: complete reversal by L-arginine. Arterioscler Thromb Vasc Biol 2002; 22:680-5. [PMID: 11950710 DOI: 10.1161/01.atv.0000013287.08141.74] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated in vivo the effect of cholesterol diet-induced hypercholesterolemia (HC) on thromboembolism in nonatherosclerotic rabbit mesenteric arterioles and venules (diameter 21 to 45 micrometer). After mechanical vessel wall injury, the ensuing thromboembolic reaction was studied by intravital videomicroscopy. A dramatic prolongation of embolization duration (median >600 seconds) was observed in the arterioles of the HC group compared with the arterioles of a normal chow-fed (NC) control group (142 seconds, P<0.0001); concomitantly, relative thrombus height increased (thrombus height/vessel diameter was 68% for the HC group and 58% for the NC group; P<0.05). By contrast, in venules, cholesterol did not affect embolization duration (42 seconds for HC group, 34 seconds for NC group) and thrombus height (66% for HC group, 64% for NC group). Furthermore, the role of endothelial NO synthesis was studied. In arterioles, stimulation of endogenous NO synthesis through mesenteric superfusion of L-arginine (1 mmol/L) completely reversed cholesterol-enhanced embolization (152 seconds) but did not influence thrombus height (63%). L-Arginine had no effect in venules of the HC group (51 seconds) and nor in the arterioles and venules of the NC group (177 seconds for arterioles, 43 seconds for venules). This study indicates that hypercholesterolemia selectively enhances thrombus formation and embolization in arterioles but not in venules and that stimulation of endogenous NO production antagonizes this enhancement of arteriolar thromboembolism.
Collapse
Affiliation(s)
- Martijn A W Broeders
- Department of Physiology, CARIM, Maastricht University, Maastricht, the Netherlands
| | | | | | | | | |
Collapse
|
217
|
Matveev SV, Smart EJ. Heterologous desensitization of EGF receptors and PDGF receptors by sequestration in caveolae. Am J Physiol Cell Physiol 2002; 282:C935-46. [PMID: 11880282 DOI: 10.1152/ajpcell.00349.2001] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) receptors have been reported to signal via caveolin-containing membranes called caveolae. In contrast, others report that EGF and PDGF receptors are exclusively associated with caveolin-devoid membranes called rafts. Our subcellular fractionation and coimmunoprecipitation studies demonstrate that, in the absence of ligand, EGF and PDGF receptors are associated with rafts. However, in the presence of ligand, EGF and PDGF receptors transiently associate with caveolae. Surprisingly, pretreatment of cells with EGF prevents PDGF-dependent phosphorylation of PDGF receptors and extracellular signal-regulated kinase (ERK) 1/2 kinase activation. Furthermore, cells pretreated with PDGF prevent EGF-dependent phosphorylation of EGF receptors and ERK1/2 kinase activation. Radioligand binding studies demonstrate that incubation of cells with EGF or PDGF causes both EGF and PDGF receptors to be reversibly sequestered from the extracellular space. Experiments with methyl-beta-cyclodextrin, filipin, and antisense caveolin-1 demonstrate that sequestration of the receptors is dependent on cholesterol and caveolin-1. We conclude that ligand-induced stimulation of EGF or PDGF receptors can cause the heterologous desensitization of the other receptor by sequestration in cholesterol-rich, caveolin-containing membranes or caveolae.
Collapse
Affiliation(s)
- Sergey V Matveev
- Department of Physiology, University of Kentucky Medical School, 800 Rose Street, Lexington, KY 40536, USA
| | | |
Collapse
|
218
|
Li XA, Titlow WB, Jackson BA, Giltiay N, Nikolova-Karakashian M, Uittenbogaard A, Smart EJ. High density lipoprotein binding to scavenger receptor, Class B, type I activates endothelial nitric-oxide synthase in a ceramide-dependent manner. J Biol Chem 2002; 277:11058-63. [PMID: 11792700 DOI: 10.1074/jbc.m110985200] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recently it has been demonstrated that high density lipoprotein (HDL) binding to scavenger receptors, class B, type I (SR-BI) stimulates endothelial nitric-oxide synthase (eNOS) activity. In the present studies we used a Chinese hamster ovary cell system and a human microvascular endothelial cell line to confirm that HDL stimulates eNOS activity in a SR-BI-dependent manner. Importantly, we have extended these studies to examine the mechanism whereby HDL binding to SR-BI stimulates eNOS. eNOS can be stimulated by an increase in intracellular calcium, by phosphorylation by Akt kinase, or by an increase in intracellular ceramide. Calcium imagining studies and experiments with the calcium chelator, 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetra(acetoxymethyl) ester demonstrated that HDL binding to SR-BI does not induce an increase in intracellular calcium. Antibodies specific for activated Akt kinase demonstrated that HDL binding to SR-BI does not induce Akt kinase activation. However, HDL binding to SR-BI caused a reversible increase in intracellular ceramide levels from 97 +/- 14 pmol/mg of protein to 501 +/- 21 pmol/mg of protein. In addition, C(2)-ceramide stimulated eNOS to the same extent as HDL, whereas C(2)-dihydroceramide did not stimulate eNOS. We conclude that HDL binding to SR-BI stimulates eNOS by increasing intracellular ceramide levels and is independent of an increase in intracellular calcium or Akt kinase phosphorylation.
Collapse
Affiliation(s)
- Xiang-An Li
- Department of Physiology, University of Kentucky Medical School, Lexington, Kentucky 40536-0230
| | | | | | | | | | | | | |
Collapse
|
219
|
Nofer JR, Kehrel B, Fobker M, Levkau B, Assmann G, von Eckardstein A. HDL and arteriosclerosis: beyond reverse cholesterol transport. Atherosclerosis 2002; 161:1-16. [PMID: 11882312 DOI: 10.1016/s0021-9150(01)00651-7] [Citation(s) in RCA: 399] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The inverse correlation between serum levels of high density lipoprotein (HDL) cholesterol and the risk of coronary heart disease, the protection of susceptible animals from atherosclerosis by transgenic manipulation of HDL metabolism, and several potentially anti-atherogenic in vitro-properties have made HDL metabolism an interesting target for pharmacological intervention in atheroslcerosis. We have previously reviewed the concept of reverse cholesterol transport, which describes both the metabolism and the classic anti-atherogenic function of HDL (Arterioscler. Thromb. Vasc. Biol. 20 2001 13). We here summarize the current understanding of additional biological, potentially anti-atherogenic properties of HDL. HDL inhibits the chemotaxis of monocytes, the adhesion of leukocytes to the endothelium, endothelial dysfunction and apoptosis, LDL oxidation, complement activation, platelet activation and factor X activation but also stimulates the proliferation of endothelial cells and smooth muscle cells, the synthesis of prostacyclin and natriuretic peptide C in endothelial cells, and the activation of proteins C and S. These anti-inflammatory, anti-oxidative, anti-aggregatory, anti-coagulant, and pro-fibrinolytic activities are exerted by different components of HDL, namley apolipoproteins, enzymes, and even specific phospholipids. This complexity further emphasizes that changes in the functionality of HDL rather than changes of plasma HDL-cholesterol levels determine the anti-atherogenicity of therapeutic alterations of HDL metabolism.
Collapse
Affiliation(s)
- Jerzy-Roch Nofer
- Institut für Klinische Chemie und Laboratoriumsmedizin, Westfälische Wilhelms-Universität, Albert Schweitzer Str. 33, 48129 Münster, Germany.
| | | | | | | | | | | |
Collapse
|
220
|
Abstract
Efflux of free cholesterol (FC) continues even when cellular FC mass is unchanged. This reflects a recirculation of preformed FC between cells and extracellular fluids which has multiple functions in cell biology including receptor recycling and signaling as well as cellular FC homeostasis. Total FC efflux is heterogeneous. Simple diffusion to mature high density lipoprotein (HDL), mainly via albumin as intermediate, initiates FC net transport driven by plasma lecithin:cholesterol acyltransferase activity. A second major efflux component reflects protein-facilitated transport from cell surface domains (caveolae, rafts) driven by FC binding to lipid-poor, pre-beta-migrating HDL (pre-beta-HDL). Facilitated efflux from caveolae, unlike simple diffusion, is highly regulated. Neither ABC1 (the protein defective in Tangier disease) nor other ATP-dependent transporters now appear likely to contribute directly to FC efflux. Their role is limited to the initial formation of a particle precursor to circulating pre-beta-HDL, which recycles without further lipid input from ATP-dependent transporter proteins. Lipid-free apolipoprotein A-I, previously considered a surrogate for pre-beta-HDL, has a reactivity much lower than that of native lipoprotein FC acceptors.
Collapse
Affiliation(s)
- C J Fielding
- Cardiovascular Research Institute and Departments of Physiology and Medicine, Box 0130, University of California Medical Center, San Francisco, CA 94143-0130, USA.
| | | |
Collapse
|
221
|
Abstract
Elevated plasma levels of HDL cholesterol or apolipoprotein A-I, the major protein moiety of HDL particles, are protective against coronary artery disease. HDL particles remove cholesterol from peripheral cells and transfer it to the liver for bile acid synthesis. The interaction between lipoproteins is not mediated through simple contact between 2 phospholipid membranes but involves specific protein-receptor interactions, charged phospholipid-phospholipid contact, and activation of cellular signaling pathways. These lead to regulation of genes or the modification of proteins involved in vasomotor function, platelet activation, thrombosis and thrombolysis, cell adhesion, apoptosis and cell proliferation, and cellular cholesterol homeostasis.
Collapse
Affiliation(s)
- B J O'Connell
- Cardiovascular Genetics Laboratory, McGill University Health Center, Royal Victoria Hospital, Montreal, Quebec, Canada
| | | |
Collapse
|
222
|
Abstract
The HDL receptor scavenger receptor class B type I plays an important role in meditating the uptake of HDL-derived cholesterol and cholesteryl ester in the liver and steroidogenic tissues. However, the mechanism by which scavenger receptor class B type I mediates selective cholesterol uptake is unclear. In hepatocytes scavenger receptor class B type I mediates the transcytosis of cholesterol into bile, appears to be expressed on both basolateral and apical membranes, and directly interacts with a PDZ domain containing protein that may modulate the activity of scavenger receptor class B type I. This suggests the involvement of scavenger receptor class B type I in higher order complexes in polarized cells. Scavenger receptor class B type I expression has been shown to alter plasma membrane cholesterol distribution and induce the formation of novel membrane structures, suggesting multiple roles for scavenger receptor class B type I in the cell. A close examination of scavenger receptor class B type I function in polarized cells may yield new insights into the mechanism of scavenger receptor class B type I-mediated HDL selective uptake and the effects of scavenger receptor class B type I on cellular cholesterol homeostasis.
Collapse
Affiliation(s)
- D L Silver
- The Division of Molecular Medicine, Department of Medicine, Columbia University, New York , NY 10032, USA.
| | | |
Collapse
|
223
|
Abstract
Increased LDL oxidation is associated with coronary artery disease. The predictive value of circulating oxidized LDL is additive to the Global Risk Assessment Score for cardiovascular risk prediction based on age, gender, total and HDL cholesterol, diabetes, hypertension, and smoking. Circulating oxidized LDL does not originate from extensive metal ion-induced oxidation in the blood but from mild oxidation in the arterial wall by cell-associated lipoxygenase and/or myeloperoxidase. Oxidized LDL induces atherosclerosis by stimulating monocyte infiltration and smooth muscle cell migration and proliferation. It contributes to atherothrombosis by inducing endothelial cell apoptosis, and thus plaque erosion, by impairing the anticoagulant balance in endothelium, stimulating tissue factor production by smooth muscle cells, and inducing apoptosis in macrophages. HDL cholesterol levels are inversely related to risk of coronary artery disease. HDL prevents atherosclerosis by reverting the stimulatory effect of oxidized LDL on monocyte infiltration. The HDL-associated enzyme paraoxonase inhibits the oxidation of LDL. PAF-acetyl hydrolase, which circulates in association with HDL and is produced in the arterial wall by macrophages, degrades bioactive oxidized phospholipids. Both enzymes actively protect hypercholesterolemic mice against atherosclerosis. Oxidized LDL inhibits these enzymes. Thus, oxidized LDL and HDL are indeed antagonists in the development of cardiovascular disease.
Collapse
MESH Headings
- 1-Alkyl-2-acetylglycerophosphocholine Esterase
- Animals
- Aryldialkylphosphatase
- Coronary Artery Disease/etiology
- Esterases/metabolism
- Humans
- Lipoproteins, HDL/antagonists & inhibitors
- Lipoproteins, HDL/physiology
- Lipoproteins, LDL/antagonists & inhibitors
- Lipoproteins, LDL/metabolism
- Lipoproteins, LDL/physiology
- Membrane Proteins
- Mice
- Models, Cardiovascular
- Phospholipases A/metabolism
- Receptors, Immunologic/biosynthesis
- Receptors, Lipoprotein
- Receptors, Scavenger
- Scavenger Receptors, Class B
- Thrombosis/etiology
Collapse
Affiliation(s)
- A Mertens
- Center for Experimental Surgery and Anesthesiology, Katholieke Universiteit Leuven, Belgium
| | | |
Collapse
|
224
|
Febbraio M, Hajjar DP, Silverstein RL. CD36: a class B scavenger receptor involved in angiogenesis, atherosclerosis, inflammation, and lipid metabolism. J Clin Invest 2001; 108:785-91. [PMID: 11560944 PMCID: PMC200943 DOI: 10.1172/jci14006] [Citation(s) in RCA: 845] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- M Febbraio
- Department of Medicine, Division of Hematology-Medical Oncology, Center of Vascular Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, New York 10021, USA.
| | | | | |
Collapse
|
225
|
Nofer JR, Levkau B, Wolinska I, Junker R, Fobker M, von Eckardstein A, Seedorf U, Assmann G. Suppression of endothelial cell apoptosis by high density lipoproteins (HDL) and HDL-associated lysosphingolipids. J Biol Chem 2001; 276:34480-5. [PMID: 11432865 DOI: 10.1074/jbc.m103782200] [Citation(s) in RCA: 264] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Apoptotic cell death following injury of vascular endothelium is assumed to play an important role in the pathogenesis of atherosclerosis. In this report, we demonstrate that high density lipoproteins (HDL), a major anti-atherogenic lipoprotein fraction, protect endothelial cells against growth factor deprivation-induced apoptosis. HDL blocked the mitochondrial pathway of apoptosis by inhibiting dissipation of mitochondrial potential (Deltapsi(m)), generation of reactive oxygen species, and release of cytochrome c into the cytoplasm. As a consequence, HDL prevented activation of caspases 9 and 3 and apoptotic alterations of the plasma membrane such as increase of permeability and translocation of phosphatidylserine. Treatment of endothelial cells with HDL induced activation of the protein kinase Akt, an ubiquitous transducer of anti-apoptotic signals, and led to phosphorylation of BAD, a major Akt substrate. Suppression of Akt activity both by wortmannin and LY-294002 or by a dominant negative Akt mutant abolished the anti-apoptotic effect of HDL. Two bioactive lysosphingolipids present in HDL particles, sphingosylphosphorylcholine and lysosulfatide, fully mimicked the survival effect of HDL by blocking the mitochondrial pathway of apoptosis and potently activating Akt. In conclusion, the present study identifies HDL as a carrier of endogenous endothelial survival factors and suggests that inhibition of endothelial apoptosis by HDL-associated lysosphingolipids may represent an important and novel aspect of the anti-atherogenic activity of HDL.
Collapse
Affiliation(s)
- J R Nofer
- Institut für Klinische Chemie und Laboratoriumsmedizin, Westfälische Wilhelms-Universität and the Institut für Arterioskleroseforschung an der Universität Münster, 48129 Münster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
226
|
Matveev S, Li X, Everson W, Smart EJ. The role of caveolae and caveolin in vesicle-dependent and vesicle-independent trafficking. Adv Drug Deliv Rev 2001; 49:237-50. [PMID: 11551397 DOI: 10.1016/s0169-409x(01)00138-7] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Caveolae can mediate endocytosis, transcytosis, and potocytosis. Our understanding of these processes as well as the elucidation of the molecular machinery involved has greatly expanded. In addition, caveolin, a 22 kDa protein often associated with caveolae, can promote the trafficking of sterol through the cytoplasm independent of vesicles. Caveolin also influences the formation, morphology, and function of caveolae. The ability of caveolae and caveolin to mediate macromolecular transport directly impacts a variety of physiological and pathophysiological processes.
Collapse
Affiliation(s)
- S Matveev
- University of Kentucky Medical School, Department of Physiology, 800 Rose Street, Lexington, KY 40536, USA
| | | | | | | |
Collapse
|
227
|
Yuhanna IS, Zhu Y, Cox BE, Hahner LD, Osborne-Lawrence S, Lu P, Marcel YL, Anderson RG, Mendelsohn ME, Hobbs HH, Shaul PW. High-density lipoprotein binding to scavenger receptor-BI activates endothelial nitric oxide synthase. Nat Med 2001; 7:853-7. [PMID: 11433352 DOI: 10.1038/89986] [Citation(s) in RCA: 564] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Atherosclerosis is the primary cause of cardiovascular disease, and the risk for atherosclerosis is inversely proportional to circulating levels of high-density lipoprotein (HDL) cholesterol. However, the mechanisms by which HDL is atheroprotective are complex and not well understood. Here we show that HDL stimulates endothelial nitric oxide synthase (eNOS) in cultured endothelial cells. In contrast, eNOS is not activated by purified forms of the major HDL apolipoproteins ApoA-I and ApoA-II or by low-density lipoprotein. Heterologous expression experiments in Chinese hamster ovary cells reveal that scavenger receptor-BI (SR-BI) mediates the effects of HDL on the enzyme. HDL activation of eNOS is demonstrable in isolated endothelial-cell caveolae where SR-BI and eNOS are colocalized, and the response in isolated plasma membranes is blocked by antibodies to ApoA-I and SR-BI, but not by antibody to ApoA-II. HDL also enhances endothelium- and nitric-oxide-dependent relaxation in aortae from wild-type mice, but not in aortae from homozygous null SR-BI knockout mice. Thus, HDL activates eNOS via SR-BI through a process that requires ApoA-I binding. The resulting increase in nitric-oxide production might be critical to the atheroprotective properties of HDL and ApoA-I.
Collapse
Affiliation(s)
- I S Yuhanna
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Abstract
Caveolae, cholesterol-rich invaginations of the plasma membrane, have been implicated as scaffolds where signaling complexes are assembled, and as portals to which recycling or newly synthesized free cholesterol is transported prior to efflux or redistribution. New data indicate that these functions may be related; membrane cholesterol content can regulate receptor-mediated signal transduction, while signals responding to membrane cholesterol levels may reach the nucleus via a parallel kinase-dependent pathway. This information from both sources may be integrated at the nuclear level to control complex biological functions such as locomotion and cell division.
Collapse
Affiliation(s)
- C J Fielding
- Cardiovascular Research Institute and Department of Physiology, University of California San Francisco, San Francisco, California 94143-0130, USA.
| |
Collapse
|
229
|
Connelly MA, de la Llera-Moya M, Monzo P, Yancey PG, Drazul D, Stoudt G, Fournier N, Klein SM, Rothblat GH, Williams DL. Analysis of chimeric receptors shows that multiple distinct functional activities of scavenger receptor, class B, type I (SR-BI), are localized to the extracellular receptor domain. Biochemistry 2001; 40:5249-59. [PMID: 11318648 DOI: 10.1021/bi002825r] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Scavenger receptor BI (SR-BI) mediates the selective uptake of high-density lipoprotein (HDL) cholesteryl ester (CE), a process by which HDL CE is taken into the cell without degradation of the HDL particle. In addition, SR-BI stimulates the bi-directional flux of free cholesterol (FC) between cells and lipoproteins, an activity that may be responsible for net cholesterol efflux from peripheral cells as well as the rapid hepatic clearance of FC from plasma HDL. SR-BI also increases cellular cholesterol mass and alters cholesterol distribution in plasma membrane domains as judged by the enhanced sensitivity of membrane cholesterol to extracellular cholesterol oxidase. In contrast, CD36, a closely related class B scavenger receptor, has none of these activities despite binding HDL with high affinity. In the present study, analyses of chimeric SR-BI/CD36 receptors and domain-deleted SR-BI have been used to test the various domains of SR-BI for functional activities related to HDL CE selective uptake, bi-directional FC flux, and the alteration of membrane cholesterol mass and distribution. The results show that each of these activities localizes to the extracellular domain of SR-BI. The N-terminal cytoplasmic tail and transmembrane domains appear to play no role in these activities other than targeting the receptor to the plasma membrane. The C-terminal tail of SR-BI is dispensable for activity as well for targeting to the plasma membrane. Thus, multiple distinct functional activities are localized to the SR-BI extracellular domain.
Collapse
Affiliation(s)
- M A Connelly
- Department of Pharmacological Sciences, University Medical Center, State University of New York at Stony Brook, 11794-8651, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Frank PG, Galbiati F, Volonte D, Razani B, Cohen DE, Marcel YL, Lisanti MP. Influence of caveolin-1 on cellular cholesterol efflux mediated by high-density lipoproteins. Am J Physiol Cell Physiol 2001; 280:C1204-14. [PMID: 11287334 DOI: 10.1152/ajpcell.2001.280.5.c1204] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Caveolin-1 is a principal structural component of caveolae membranes. These membrane microdomains participate in the regulation of signaling, transcytosis, and cholesterol homeostasis at the plasma membrane. In the present study, we determined the effect of caveolin-1 expression on cellular cholesterol efflux mediated by high-density lipoprotein (HDL). We evaluated this effect in parental NIH/3T3 cells as well as in two transformed NIH/3T3 cell lines in which caveolin-1 protein levels are dramatically downregulated. Compared with parental NIH/3T3 cells, these two transformed cell lines effluxed cholesterol more rapidly to HDL. In addition, NIH/3T3 cells harboring caveolin-1 antisense also effluxed cholesterol more rapidly to HDL. However, this effect was not due to changes in total cellular cholesterol content. We further showed that chronic HDL exposure reduced caveolin-1 protein expression in NIH/3T3 cells. HDL exposure also inhibited caveolin-1 promoter activity, suggesting a direct negative effect of HDL on caveolin-1 gene transcription. Moreover, we showed that HDL-induced downregulation of caveolin-1 prevents the uptake of oxidized low-density lipoprotein in human endothelial cells. These data suggest a novel proatherogenic role for caveolin-1, i.e., regarding the uptake and/or transcytosis of modified lipoproteins.
Collapse
Affiliation(s)
- P G Frank
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | |
Collapse
|
231
|
Nuszkowski A, Gräbner R, Marsche G, Unbehaun A, Malle E, Heller R. Hypochlorite-modified low density lipoprotein inhibits nitric oxide synthesis in endothelial cells via an intracellular dislocalization of endothelial nitric-oxide synthase. J Biol Chem 2001; 276:14212-21. [PMID: 11278358 DOI: 10.1074/jbc.m007659200] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hypochlorous acid/hypochlorite, generated by the myeloperoxidase/H(2)O(2)/halide system of activated phagocytes, has been shown to oxidize/modify low density lipoprotein (LDL) in vitro and may be involved in the formation of atherogenic lipoproteins in vivo. Accordingly, hypochlorite-modified (lipo)proteins have been detected in human atherosclerotic lesions where they colocalize with macrophages and endothelial cells. The present study investigates the influence of hypochlorite-modified LDL on endothelial synthesis of nitric oxide (NO) measured as formation of citrulline (coproduct of NO) and cGMP (product of the NO-activated soluble guanylate cyclase) upon cell stimulation with thrombin or ionomycin. Pretreatment of human umbilical vein endothelial cells with hypochlorite-modified LDL led to a time- and concentration-dependent inhibition of agonist-induced citrulline and cGMP synthesis compared with preincubation of cells with native LDL. This inhibition was neither due to a decreased expression of endothelial NO synthase (eNOS) nor to a deficiency of its cofactor tetrahydrobiopterin. Likewise, the uptake of l-arginine, the substrate of eNOS, into the cells was not affected. Hypochlorite-modified LDL caused remarkable changes of intracellular eNOS distribution including translocation from the plasma membrane and disintegration of the Golgi location without altering myristoylation or palmitoylation of the enzyme. In contrast, cyclodextrin known to deplete plasma membrane of cholesterol and to disrupt caveolae induced only a disappearance of eNOS from the plasma membrane that was not associated with decreased agonist-induced citrulline and cGMP formation. The present findings suggest that mislocalization of NOS accounts for the reduced NO formation in human umbilical vein endothelial cells treated with hypochlorite-modified LDL and point to an important role of Golgi-located NOS in these processes. We conclude that inhibition of NO synthesis by hypochlorite-modified LDL may be an important mechanism in the development of endothelial dysfunction and early pathogenesis of atherosclerosis.
Collapse
MESH Headings
- Antioxidants/pharmacology
- Arginine/pharmacokinetics
- Biopterins/analogs & derivatives
- Biopterins/biosynthesis
- Blotting, Western
- Cell Membrane/metabolism
- Cells, Cultured
- Centrifugation, Density Gradient
- Citrulline/biosynthesis
- Cyclic GMP/metabolism
- Cyclodextrins/pharmacology
- Dose-Response Relationship, Drug
- Electrophoresis, Polyacrylamide Gel
- Endothelium, Vascular/cytology
- Endothelium, Vascular/enzymology
- Golgi Apparatus/metabolism
- Humans
- Hypochlorous Acid/metabolism
- Immunoblotting
- Immunohistochemistry
- Ionomycin/pharmacology
- Ionophores/pharmacology
- Lipoproteins, LDL/metabolism
- Microscopy, Fluorescence
- Myristic Acids/metabolism
- Nitric Oxide/biosynthesis
- Nitric Oxide Synthase/metabolism
- Nitric Oxide Synthase Type III
- Palmitic Acids/metabolism
- Precipitin Tests
- RNA, Messenger/metabolism
- Subcellular Fractions/metabolism
- Sucrose/metabolism
- Time Factors
- Umbilical Veins/cytology
Collapse
Affiliation(s)
- A Nuszkowski
- Center of Vascular Biology and Medicine, Friedrich-Schiller-University of Jena, 99089 Erfurt, Germany
| | | | | | | | | | | |
Collapse
|
232
|
Marsche G, Levak-Frank S, Quehenberger O, Heller R, Sattler W, Malle E. Identification of the human analog of SR‐BI and LOX‐1 as receptors for hypochlorite‐modified high‐density lipoprotein on human umbilical venous endothelial cells. FASEB J 2001. [DOI: 10.1096/fsb2fj000532fje] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Gunther Marsche
- Karl-Franzens University, Institute of Medical BiochemistryHarrachgasse 21, A-8010 Graz Austria
| | - Sanja Levak-Frank
- Karl-Franzens University, Institute of Medical BiochemistryHarrachgasse 21, A-8010 Graz Austria
| | | | - Regine Heller
- Center of Vascular Biology and Medicine Friedrich-Schiller-University Erfurt Germany
| | - Wolfgang Sattler
- Karl-Franzens University, Institute of Medical BiochemistryHarrachgasse 21, A-8010 Graz Austria
| | - Ernst Malle
- Karl-Franzens University, Institute of Medical BiochemistryHarrachgasse 21, A-8010 Graz Austria
| |
Collapse
|
233
|
Abstract
Endothelial dysfunction has been shown in a wide range of vascular disorders including atherosclerosis and related diseases. Here, we examine and address the complex relationship among nitric oxide (NO)-mediated pathways and atherogenesis. In view of the numerous pathophysiological actions of NO, abnormalities could potentially occur at many sites: (a) impairment of membrane receptors in the arterial wall that interact with agonists or physiological stimuli capable of generating NO; (b) reduced concentrations or impaired utilization of l-arginine; (c) reduction in concentration or activity both of inducible and endothelial NO synthase; (d) impaired release of NO from the atherosclerotic damaged endothelium; (e) impaired NO diffusion from endothelium to vascular smooth muscle cells followed by decreased sensitivity to its vasodilator action; (f) local enhanced degradation of NO by increased generation of free radicals and/or oxidation-sensitive mechanisms; and (g) impaired interaction of NO with guanylate cyclase and consequent limitation of cyclic GMP production. Therefore, one target for new drugs should be the preservation or restoration of NO-mediated signaling pathways in arteries. Such novel therapeutic strategies may include administration of l-arginine/antioxidants and gene-transfer approaches.
Collapse
Affiliation(s)
- C Napoli
- Department of Medicine, Federico II University of Naples, Naples, Italy.
| | | |
Collapse
|
234
|
Reaven E, Leers-Sucheta S, Nomoto A, Azhar S. Expression of scavenger receptor class B type 1 (SR-BI) promotes microvillar channel formation and selective cholesteryl ester transport in a heterologous reconstituted system. Proc Natl Acad Sci U S A 2001; 98:1613-8. [PMID: 11171999 PMCID: PMC29305 DOI: 10.1073/pnas.98.4.1613] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In the "selective" cholesteryl ester (CE) uptake process, surface-associated lipoproteins [high density lipoprotein (HDL) and low density lipoprotein] are trapped in the space formed between closely apposed surface microvilli (microvillar channels) in hormone-stimulated steroidogenic cells. This is the same location where an HDL receptor (SR-BI) is found. In the current study, we sought to understand the relationship between SR-BI and selective CE uptake in a heterologous insect cell system. Sf9 (Spodoptera frugiperda) cells overexpressing recombinant SR-BI were examined for (i) SR-BI protein by Western blot analysis and light or electron immunomicroscopy, and (ii) selective lipoprotein CE uptake by the use of radiolabeled or fluorescent (BODIPY-CE)-labeled HDL. Noninfected or infected control Sf9 cells do not express SR-BI, show microvillar channels, or internalize CEs. An unexpected finding was the induction of a complex channel system in Sf9 cells expressing SR-BI. SR-BI-expressing cells showed many cell surface double-membraned channels, immunogold SR-BI, apolipoprotein (HDL) labeling of the channels, and high levels of selective HDL-CE uptake. Thus, double-membraned channels can be induced by expression of recombinant SR-BI in a heterologous system, and these specialized structures facilitate both the binding of HDL and selective HDL-CE uptake.
Collapse
MESH Headings
- Animals
- Biological Transport
- Blotting, Western/methods
- Boron Compounds
- CD36 Antigens/biosynthesis
- CD36 Antigens/genetics
- Cell Line
- Cholesterol Esters/metabolism
- Fluorescent Dyes
- Iodine Radioisotopes
- Isotope Labeling
- Lipoproteins, HDL/metabolism
- Lipoproteins, HDL3
- Membrane Proteins
- Microscopy, Electron/methods
- Microscopy, Fluorescence/methods
- Microvilli/metabolism
- Protein Binding
- Rats
- Receptors, Immunologic
- Receptors, Lipoprotein/biosynthesis
- Receptors, Lipoprotein/genetics
- Receptors, Scavenger
- Scavenger Receptors, Class B
- Spodoptera
- Tritium
Collapse
Affiliation(s)
- E Reaven
- Geriatric Research, Education, and Clinical Center, Department of Veterans Affairs, Palo Alto Health Care System, Palo Alto, CA 94304, USA.
| | | | | | | |
Collapse
|
235
|
Fielding CJ, Fielding PE. Cholesterol and caveolae: structural and functional relationships. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1529:210-22. [PMID: 11111090 DOI: 10.1016/s1388-1981(00)00150-5] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Caveolae are free cholesterol (FC)- and sphingolipid-rich surface microdomains abundant in most peripheral cells. Caveolin, a FC binding protein, is a major structural element of these domains. Caveolae serve as portals to regulate cellular FC homeostasis, possibly via their association with ancillary proteins including scavenger receptor B1. The FC content of caveolae regulates the transmission of both extracellular receptor-mediated and endogenous signal transduction via changes in the composition of caveolin-associated complexes of signaling intermediates. By controlling surface FC content, reporting membrane changes by signal transduction to the nucleus, and regulating signal traffic in response to extracellular stimuli, caveolae exert a multifaceted influence on cell physiology including growth and cell division, adhesion, and hormonal response. Cell surface lipid 'rafts' may assume many of the functions of caveolae in cells with low levels of caveolin.
Collapse
Affiliation(s)
- C J Fielding
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, CA 94143, USA.
| | | |
Collapse
|
236
|
Chambliss KL, Yuhanna IS, Mineo C, Liu P, German Z, Sherman TS, Mendelsohn ME, Anderson RG, Shaul PW. Estrogen receptor alpha and endothelial nitric oxide synthase are organized into a functional signaling module in caveolae. Circ Res 2000; 87:E44-52. [PMID: 11090554 DOI: 10.1161/01.res.87.11.e44] [Citation(s) in RCA: 283] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Estrogen causes nitric oxide (NO)-dependent vasodilation due to estrogen receptor (ER) alpha-mediated, nongenomic activation of endothelial NO synthase (eNOS). The subcellular site of interaction between ERalpha and eNOS was determined in studies of isolated endothelial cell plasma membranes. Estradiol (E(2), 10(-8) mol/L) caused an increase in eNOS activity in plasma membranes in the absence of added calcium, calmodulin, or eNOS cofactors, which was blocked by ICI 182,780 and ERalpha antibody. Immunoidentification studies detected the same 67-kDa protein in endothelial cell nucleus, cytosol, and plasma membrane. Plasma membranes from COS-7 cells expressing eNOS and ERalpha displayed ER-mediated eNOS stimulation, whereas membranes from cells expressing eNOS alone or ERalpha plus a myristoylation-deficient mutant eNOS were insensitive. Fractionation of endothelial cell plasma membranes revealed ERalpha protein in caveolae, and E(2) caused stimulation of eNOS in isolated caveolae that was ER-dependent; noncaveolae membranes were insensitive. Acetylcholine and bradykinin also activated eNOS in isolated caveolae. Furthermore, the effect of E(2) on eNOS in caveolae was prevented by calcium chelation. Thus, a subpopulation of ERalpha is localized to endothelial cell caveolae where they are coupled to eNOS in a functional signaling module that may regulate the local calcium environment. The full text of this article is available at http://www.circresaha.org.
Collapse
Affiliation(s)
- K L Chambliss
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75235-9063, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Abstract
CD36 has been associated with diverse normal and pathologic processes. These include scavenger receptor functions (uptake of apoptotic cells and modified lipid), lipid metabolism and fatty acid transport, adhesion, angiogenesis, modulation of inflammation, transforming growth factor-beta activation, atherosclerosis, diabetes and cardiomyopathy. Although CD36 was identified more than 25 years ago, it is only with the advent of recent genetic technology that in-vivo evidence has emerged for its physiologic and pathologic relevance. As these in-vivo studies are expanded, we will gain further insight into the mechanism(s) by which CD36 transmits a cellular signal, and this will allow the design of specific therapeutics that impact on a particular function of CD36.
Collapse
Affiliation(s)
- R L Silverstein
- Weill Medical College of Cornell University, Department of Medicine, New York, New York 10021, USA
| | | |
Collapse
|